Entry - *600112 - DYNEIN, CYTOPLASMIC 1, HEAVY CHAIN 1; DYNC1H1 - OMIM
* 600112

DYNEIN, CYTOPLASMIC 1, HEAVY CHAIN 1; DYNC1H1


Alternative titles; symbols

DNCH1
DYNEIN, CYTOPLASMIC-LIKE; DNCL; DNECL
DYNEIN HEAVY POLYPEPTIDE, CYTOPLASMIC; DHC1; DHC1A
DNCHC1


HGNC Approved Gene Symbol: DYNC1H1

Cytogenetic location: 14q32.31     Genomic coordinates (GRCh38): 14:101,964,573-102,056,443 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
14q32.31 Charcot-Marie-Tooth disease, axonal, type 2O 614228 AD 3
Cortical dysplasia, complex, with other brain malformations 13 614563 AD 3
Spinal muscular atrophy, lower extremity-predominant 1, AD 158600 AD 3

TEXT

Description

The DYNC1H1 gene encodes a large (over 530 kD) crucial subunit of the cytoplasmic dynein complex (summary by Poirier et al., 2013). Dyneins are a group of microtubule-activated ATPases that serve to convert chemical energy into mechanical energy. They have been divided into 2 large subgroups, namely, the axonemal and cytoplasmic dyneins. Cytoplasmic dynein has been implicated in a variety of other forms of intracellular motility, including retrograde axonal transport, protein sorting between apical and basolateral surfaces, and redistribution of organelles like endosomes and lysosomes. Molecules of conventional cytoplasmic dynein contain 2 heavy chain polypeptides and a number of intermediate and light chains. They sediment at approximately 20S (Vaisberg et al., 1993).


Cloning and Expression

Mikami et al. (1993) isolated cDNAs encoding the heavy chain of MAP1C, a rat cytoplasmic dynein. The predicted 4,644-amino acid protein contains 4 ATP-binding consensus sequences. Southern blot analysis suggested that there is a single cytoplasmic dynein gene in rat. Gibbons et al. (1994) identified DYH1a, a sea urchin cytoplasmic dynein with homology to MAP1C. Criswell et al. (1996) reported that MAP1C, or DHC1a, expression was unchanged during ciliogenesis in primary rat tracheal epithelial cells.

By screening an adenocarcinoma cell line library with p22, Vaisberg et al. (1996) isolated a DHC1 cDNA. The predicted partial protein sequence shares 99% and 34% identity with rat DHC1a and human DNHC2 (603297), respectively. Antibodies against DHC1 recognized a high molecular mass protein on Western blots of extracts from several mammalian cell lines. Northern blot analysis revealed that DHC1 is expressed as an approximately 15-kb mRNA in several mammalian cells lines and human tissues, including those that make neither cilia nor flagella. By immunofluorescence, Vaisberg et al. (1996) found that DHC1 localizes in a punctate pattern in the cytoplasm that is generally brighter in the perinuclear area and dimmer near the cell periphery. DHC1 redistributes during mitosis to the kinetochores and mitotic spindle.


Gene Function

Vaisberg et al. (1993) cloned a partial cDNA (p22) encoding the putative ATP-hydrolytic site of the human conventional cytoplasmic dynein heavy chain (DHC). Antibodies against the resulting polypeptide inhibited dynein motor activity in vitro. Injection of these antibodies into mitotic mammalian cells blocked the formation of spindles in prophase or during recovery from nocodazole treatment at later stages of mitosis. The cells became arrested with unseparated centrosomes and formed monopolar spindles. However, there was no detectable effect on chromosome attachment to a bipolar spindle or on chromosome movements during anaphase. Vaisberg et al. (1993) suggested that cytoplasmic dynein plays a unique and important role in the initial events of bipolar spindle formation, while any later roles in mitosis may be redundant.

Sasaki et al. (2000) demonstrated that Lis1 (PAFAH1B1; 601545) directly interacts with Dync1h1 and Ndel1 (607538) in the developing mouse brain. Lis1 and Ndel1 colocalized predominantly at the centrosome in early neuroblasts, but redistributed to axons in association with retrograde dynein motor proteins. Ndel1 and Lis1 regulated the distribution of Dync1h1 along microtubules, establishing a direct functional link between LIS1, NDEL1, and microtubule motors during neuronal migration and axonal retrograde transport in the mammalian brain.

Kural et al. (2005) used fluorescence imaging with 1-nanometer accuracy (FIONA) to analyze organelle movement by conventional kinesin (602809) and cytoplasmic dynein in a cell. They located a green fluorescent protein (GFP)-tagged peroxisome in cultured Drosophila S2 cells to within 1.5 nanometers in 1.1 milliseconds, a 400-fold improvement in temporal resolution, sufficient to determine the average step size to be approximately 8 nanometers for both dynein and kinesin. Furthermore, Kural et al. (2005) found that dynein and kinesin do not work against each other in vivo during peroxisome transport. Rather, multiple kinesins or multiple dyneins work together, producing up to 10 times the in vitro speed.

To determine the effects of tau (157140) on dynein and kinesin motility, Dixit et al. (2008) conducted single-molecule studies of motor proteins moving along tau-decorated microtubules. Dynein tended to reverse direction, whereas kinesin tended to detach at patches of bound tau. Kinesin was inhibited at about a tenth of the tau concentration that inhibited dynein, and the microtubule-binding domain of tau was sufficient to inhibit motor activity. The differential modulation of dynein and kinesin motility suggested that microtubule-associated proteins (MAPs) can spatially regulate the balance of microtubule-dependent axonal transport.


Mapping

By fluorescence in situ hybridization, Narayan et al. (1994) localized the heavy chain gene of human cytoplasmic dynein to the terminal region of 14q. Indirect confirmation of this result was provided by the fact that the mouse cytoplasmic dynein heavy chain had been localized to murine chromosome 12, which shows extensive homology of synteny with human 14 (Mikami et al., 1993).

By genomic sequence analysis, Pazour et al. (2006) mapped the DNCH1 gene to chromosome 14q32.


Biochemical Features

Crystal Structure

Carter et al. (2008) reported the crystal structure of the mouse cytoplasmic dynein microtubule binding domain (MTBD) and a portion of the coiled coil, which supports a mechanism by which the ATPase domain and the MTBD may communicate through a shift in the heptad registry of the coiled coil. Surprisingly, functional data suggested that the MTBD and not the ATPase domain is the main determinant of the direction of dynein motility.

Urnavicius et al. (2018) used electron microscopy and single-molecule studies to show that adaptors can recruit a second dynein to dynactin (601143). Whereas BICD2 (609797) is biased towards recruiting a single dynein, the adaptors BICDR1 (617002) and HOOK3 (607825) predominantly recruit 2 dyneins. Urnavicius et al. (2018) found that the shift towards a double dynein complex increases both the force and speed of the microtubule motor. The 3.5-angstrom resolution cryoelectron microscopy reconstruction of a dynein tail-dynactin-BICDR1 complex revealed how dynactin can act as a scaffold to coordinate 2 dyneins side by side. Urnavicius et al. (2018) concluded that their work provided a structural basis for understanding how diverse adaptors recruit different numbers of dyneins and regulate the motile properties of the dynein-dynactin transport machine.


Molecular Genetics

Charcot-Marie-Tooth Disease, Axonal, Type 2O

In affected members of a large 4-generation family with autosomal dominant axonal Charcot-Marie-Tooth disease type 2O (CMT2O; 614228), Weedon et al. (2011) identified a heterozygous mutation in the DYNC1H1 gene (H306R; 600112.0001). The mutation was identified by exome sequencing. Affected individuals had onset in childhood of delayed motor milestones and abnormal gait and falls associated with distal lower limb weakness and wasting and distal sensory impairment. Weedon et al. (2011) noted that mouse models had implicated mutations in this gene in neuropathic disease.

Complex Cortical Dysplasia With Other Brain Malformations 13

By family-based exome sequencing of 10 case-parent trios with global developmental delay, Vissers et al. (2010) identified a de novo heterozygous mutation in the DYNC1H1 gene (H3822P; 600112.0002) in 1 patient. He had hypotonia and mild dysmorphic facial features. Follow-up of the patient at age 6 years by Willemsen et al. (2012) noted that he had hypotonia, hyporeflexia, and broad-based waddling gait with toe-walking. Reevaluation of brain MRI showed complex cortical dysplasia with other brain malformations (CDCBM13; 614563). Willemsen et al. (2012) identified a second de novo heterozygous mutation in the DYNC1H1 gene (E1518K; 600112.0003) in a 51-year-old woman with severe developmental delay since infancy and an inability to walk or speak. She had mild dysmorphic features, seizures, and spastic tetraplegia. Cerebral CT scan at the age 46 years showed enlarged ventricles and clear signs of cortical malformation with wide opercular regions and an abnormal flat cortex with only a few simple and shallow sulci; MRI scan was not possible. Willemsen et al. (2012) noted that DYNC1H1 interacts with LIS1 (601545), haploinsufficiency of which results in the severe neuronal migration disorder lissencephaly-1 (607432), and that Dync1h1 mutant mice show neuronal migration defects (Ori-McKenney and Vallee, 2011), providing evidence of the pathogenicity of the mutations. Willemsen et al. (2012) also noted that their 2 patients showed variable signs consistent with peripheral neuropathy and that some of the patients with CMT2O (Weedon et al., 2011) showed learning difficulties, indicating that DYNC1H1 mutations may result in a broad neurologic phenotypic spectrum.

Poirier et al. (2013) identified 8 different de novo heterozygous mutations in the DYNC1H1 gene (see, e.g., 600112.0007-600112.0009) in 8 unrelated patients ascertained for evaluation due to malformations of cortical development (CDCBM13). The patients had global developmental delay, and most had early-onset seizures. Mutations in the first several patients were found by whole-exome sequencing, whereas subsequent patients were identified by direct sequencing of this gene in a larger cohort of affected individuals. In vitro functional expression studies of 2 of the variants showed that the mutant proteins had decreased microtubule binding affinity compared to wildtype. In addition, there was 1 family in which a mother and her 2 children carried a missense variant (K3241T): 1 of the children had mild intellectual disability, but the mother and the other child had normal cognition. All 3 were normocephalic, showed posterior pachygyria, and had focal seizures. No functional studies were performed on the K3241T variant, which occurred at a nonconserved residue.

Jamuar et al. (2014) used a customized panel of known and candidate genes associated with brain malformations to apply targeted high-coverage sequencing (depth greater than or equal to 200x) to leukocyte-derived DNA samples from 158 individuals with brain malformations. They found potentially causal mutations in the candidate gene DYNC1H1 in 2 individuals with pachygyria; in a parallel study they found de novo mutations in DYNC1H1 in 2 other individuals with pachygyria. The 4 individuals had strikingly similar MRI findings, with posterior-predominant pachygyria, thickened cortex in the perisylvian region, and mildly dysmorphic corpus callosum (CDCBM13).

Lower Extremity-Predominant Spinal Muscular Atrophy 1, Autosomal Dominant

In affected members of a large family with autosomal dominant lower extremity-predominant spinal muscular atrophy-1 (SMALED1; 158600) originally reported by Harms et al. (2010), Harms et al. (2012) identified a heterozygous mutation in the DYNC1H1 gene (I584L; 600112.0004). Patient skin fibroblasts showed normal binding to microtubules in the absence of ATP, but markedly decreased binding to microtubules in the presence of ATP. The mutant dynein also appeared to disrupt the stability of the dynein complex. Two additional families with a similar disorder were found to carry heterozygous DYNC1H1 mutations (600112.0005 and 600112.0006). The findings were similar to those observed in Loa homozygous mice (Hafezparast et al., 2003).

In 2 Japanese sibs with autosomal dominant lower extremity spinal muscular atrophy and no sensory symptoms, Tsurusaki et al. (2012) identified a heterozygous missense mutation in the DYNC1H1 gene (H306R; 600112.0001). The mutation, which was found by exome sequencing, was inherited from their mother, who had mild symptoms. The same mutation had previously been found by Weedon et al. (2011) in a family with autosomal dominant axonal Charcot-Marie-Tooth disease type 2O (CMT2O; 614228).

Based on findings in mutant mice, Eschbach et al. (2013) studied the mitochondria in fibroblasts derived from SMALED1 patients with the K671E (600112.0005) and I584L mutations. Both cell lines showed intensely fragmented mitochondria, with smaller mitochondria in K671E cells and increased areas of individual mitochondria in I584L cells. Both cell lines also showed decreased levels of mitofusin-1 (MFN1; 608506). Eschbach et al. (2013) suggested that dysfunction of mitochondrial transport may contribute to disease pathogenesis in patients with DYNC1H1 mutations.


Animal Model

Harada et al. (1998) generated Dnchc1 knockout mice by targeted disruption. No embryos were identified at 8.5 days postcoitum. Cultured blastocysts of Dnchc1-null embryos demonstrated a highly vesiculated Golgi complex that was distributed throughout the cytoplasm. Endosomes and lysosomes were not concentrated near the nucleus but were distributed evenly throughout the cytoplasm.

'Legs at odd angles' (Loa) and 'Cramping 1' (cra1) are mouse phenotypes that arose from ENU (N-ethyl-N-nitrosourea) mutagenesis. They are transmitted as autosomal dominant traits and give rise to age-related progressive loss of muscle tone and locomotor ability in heterozygous mice without a major reduction in life span. Homozygous mice show a more severe phenotype with an inability to feed and move, and die within 24 hours of birth. Hafezparast et al. (2003) showed that Loa is caused by a T-to-A transversion in the Dnchc1 gene that changes the phenylalanine at codon 580 to a tyrosine. The Cra1 phenotype results from an A-to-G transition leading to a tyrosine-to-cysteine substitution at codon 1055 of the Dnchc1 gene. Intercrossing heterozygous Cra1/+ with Loa/+ heterozygous mice yielded compound heterozygotes that died shortly after birth, demonstrating that the phenotypes are allelic. Homozygous and compound heterozygous mice developed Lewy-like inclusion bodies, and homozygous mice displayed significant loss of spinal anterior horn cells in utero. The Loa and Cra1 mutations do not cause overt deficits across the range of known DNCHC1 functions but result in a specific defect in fast retrograde transport that appears to manifest only in alpha motor neurons. Hafezparast et al. (2003) concluded that the Loa and Cra1 mutations exhibit remarkable similarities to specific features of human pathology for amyotrophic lateral sclerosis (ALS; 105400) and other motor neuron degeneration phenotypes including Lewy body-like inclusions containing SOD1 (147450), CDK5 (123831), neurofilaments, and ubiquitin (191339).

Braunstein et al. (2010) studied the 'cramping' mouse (Cra), which is heterozygous for a tyr1055-to-cys mutation in Dync1h1 that impairs retrograde axonal transport. These mice exhibited motor and behavioral abnormalities including hindlimb clasping, early muscle weakness, incoordination, and hyperactivity. In vivo brain imaging using magnetic resonance imaging showed striatal atrophy and lateral ventricle enlargement. In the striatum, altered dopamine signaling, decreased dopamine D1 receptor (DRD1; 126449) and D2 receptor (DRD2; 126450) binding in positron emission tomography SCAN, and prominent astrocytosis were observed, although there was no neuronal loss either in the striatum or substantia nigra. In vitro, dynein mutant striatal neurons displayed strongly impaired neuritic morphology. The authors concluded that dynein is required for the normal morphology and function of striatal neurons, and may play a role in the pathogenesis of neurodegenerative disorders of the basal ganglia such as Perry syndrome (168605) and Huntington disease (HD; 143100).

Eschbach et al. (2013) found that cultured embryonic fibroblasts from Cra1 mice had profoundly disrupted mitochondrial networks, including fragmented mitochondria and mitochondrial aggregates, associated with decreased mitofusin-1 (MFN1; 608506). Skeletal muscle from these mice showed progressive mitochondrial dysfunction with decreased respiration and altered energy metabolism. In addition, mutant mice developed late-onset glucose intolerance consistent with mitochondrial dysfunction. Dync1h1 mutant fibroblasts showed impaired perinuclear clustering of mitochondria in response to mitochondrial uncoupling. These findings indicated that dynein function is required for the maintenance of mitochondrial morphology and function.

Ori-McKenney and Vallee (2011) demonstrated that Loa homozygous mice had defects in neocortical lamination and neuronal migration resulting from a reduction in the rate of radial migration of bipolar neurons. Examination of brains from mutant mice showed blurred laminar boundaries, indicating cortical disorganization. The hippocampal dentate gyrus was smaller than wildtype. There was also a decrease in axonal extension within the brain, indicating that dynein processivity is necessary for axon elongation. The findings were similar, but less severe, than those reported in Lis1 (601545) compound heterozygous mutant mice.


ALLELIC VARIANTS ( 9 Selected Examples):

.0001 CHARCOT-MARIE-TOOTH DISEASE, AXONAL, TYPE 2O

SPINAL MUSCULAR ATROPHY, LOWER EXTREMITY-PREDOMINANT, 1, AUTOSOMAL DOMINANT, INCLUDED
DYNC1H1, HIS306ARG
  
RCV000022929...

Charcot-Marie-Tooth Disease, Axonal, Type 2O

In affected members of a large 4-generation family with autosomal dominant axonal Charcot-Marie-Tooth disease type 2O (CMT2O; 614228), Weedon et al. (2011) identified a heterozygous 917A-G transition in the DYNC1H1 gene, resulting in a his306-to-arg (H306R) substitution at a highly conserved residue in the homodimerization domain. Affected individuals had onset in childhood of delayed motor milestones and abnormal gait and falls associated with distal lower limb weakness and wasting and distal sensory impairment. Weedon et al. (2011) noted that mouse models had implicated mutations in this gene in neuropathic disease.

Spinal Muscular Atrophy, Lower Extremity-Predominant 1, Autosomal Dominant

Tsurusaki et al. (2012) identified a heterozygous H306R mutation in the DYNC1H1 gene in 2 Japanese sibs with autosomal dominant lower extremity-predominant spinal muscular atrophy-1 (SMALED1; 158600) without sensory impairment. The mother, who had mild symptoms, also carried the mutation. The mutation, which was found by exome sequencing and confirmed by Sanger sequencing, was not present in the dbSNP or 1000 Genomes databases, in 33 in-house exomes, or in 177 Japanese control individuals.


.0002 CORTICAL DYSPLASIA, COMPLEX, WITH OTHER BRAIN MALFORMATIONS 13

DYNC1H1, HIS3822PRO
  
RCV000022930...

In a 4-year-old boy with complex cortical dysplasia with other brain malformations-13 (CDCBM13; 614563), Vissers et al. (2010) identified a de novo heterozygous c.11465A-C transversion (c.11465A-C, NM_001376) in the DYNC1H1 gene, resulting in a his3822-to-pro (H3822P) substitution at a highly conserved residue in the stem domain of the protein. The mutation was found by family-based exome sequencing, and was not found in 1,664 control chromosomes. The patient showed hypotonia at age 6 months, followed by delayed psychomotor development. Mild dysmorphic features included prominent forehead, plagiocephaly, hypotonic face with downslanting palpebral fissures, and short, broad hands and feet. Brain MRI was reported as normal. Follow-up of the patient at age 6 years by Willemsen et al. (2012) noted that he had hypotonia, hyporeflexia, and broad-based waddling gait with toe-walking. Reevaluation of brain MRI showed signs of bilateral cortical malformation with deficient gyration of the frontal lobes and an area suggestive of focal cortical dysplasia, consistent with a neuronal migration defect.


.0003 CORTICAL DYSPLASIA, COMPLEX, WITH OTHER BRAIN MALFORMATIONS 13

DYNC1H1, GLU1518LYS
  
RCV000022931...

In a 51-year-old woman with complex cortical dysplasia with other brain malformations-13 (CDCBM13; 614563), Willemsen et al. (2012) identified a de novo heterozygous c.4552G-A transition (c.4552G-A, NM_001376.4) in the DYNC1H1 gene, resulting in a glu1518-to-lys (E1518K) substitution at a highly conserved residue in the motor domain of the protein. The mutation was not found in 445 control exomes. She had severe developmental delay with an inability to walk or speak and generalized seizures since age 3 years. Craniofacial features included brachycephaly, prominent forehead, hypertelorism, deep-set eyes, wide mouth with everted lower lip, and downturned corners of the mouth. Other features included short stature, microcephaly, clubfeet, small hands and feet with short toes, kyphoscoliosis, spastic tetraplegia, and swallowing difficulties. Cerebral CT scan at the age 46 years showed enlarged ventricles and clear signs of cortical malformation with wide opercular regions and an abnormal flat cortex with only a few simple and shallow sulci; MRI scan was not possible.


.0004 SPINAL MUSCULAR ATROPHY, LOWER EXTREMITY-PREDOMINANT, 1, AUTOSOMAL DOMINANT

DYNC1H1, ILE584LEU
  
RCV000022932...

In affected members of a large family with autosomal dominant lower extremity-predominant spinal muscular atrophy (SMALED1; 158600) originally reported by Harms et al. (2010), Harms et al. (2012) identified a heterozygous 1750A-C transversion in exon 8 of the DYNC1H1 gene, resulting in an ile584-to-leu (I584L) substitution at a highly conserved residue in the tail domain of the dynein heavy chain, a highly conserved region critical for organizing multiple dynein subunits into a complex. The mutation was not found in 500 controls or the 1000 Genomes Project. Affected individuals had early-childhood onset of proximal leg weakness with muscle atrophy and nonlength-dependent motor neuron disease without sensory involvement. Patient skin fibroblasts showed normal binding to microtubules in the absence of ATP, but markedly decreased binding to microtubules in the presence of ATP. The mutant dynein also appeared to disrupt the stability of the dynein complex. The findings were similar to those observed in Loa homozygous mice (Hafezparast et al., 2003; Ori-McKenney et al., 2010).


.0005 SPINAL MUSCULAR ATROPHY, LOWER EXTREMITY-PREDOMINANT, 1, AUTOSOMAL DOMINANT

DYNC1H1, LYS671GLU
  
RCV000022933...

In affected members of a 3-generation family with autosomal dominant lower extremity-predominant spinal muscular atrophy (SMALED1; 158600), Harms et al. (2012) identified a heterozygous 2011A-G transition in exon 8 of the DYNC1H1 gene, resulting in a lys671-to-glu (K671E) substitution at a highly conserved residue in the tail domain of the dynein heavy chain. The mutation was not found in 500 controls or the 1000 Genomes Project. The 3 affected individuals showed waddling gait from early childhood, with awkward running due to lower limb weakness; upper limbs were not affected. Muscle atrophy and weakness confined to the lower limbs showed little progression throughout life. There was a notable strength discrepancy between knee extension and flexion, with the quadriceps showing significant weakness. Deep tendon reflexes were reduced at the knees, but normal elsewhere. Nerve conduction studies showed small motor responses and normal sensory responses; EMG showed chronic denervation. One patient had heel cord contractures and inturning feet, whereas another had fasciculations of the calves.


.0006 SPINAL MUSCULAR ATROPHY, LOWER EXTREMITY-PREDOMINANT, 1, AUTOSOMAL DOMINANT

DYNC1H1, TYR970CYS
  
RCV000022934...

In a 3.5-year-old girl with autosomal dominant lower extremity-predominant spinal muscular atrophy (SMALED1; 158600), Harms et al. (2012) identified a heterozygous 3170A-G transition in exon 11 of the DYNC1H1 gene, resulting in a tyr970-to-cys (Y970C) substitution at a highly conserved residue. The mutation was not found in 500 controls or the 1000 Genomes Project. The patient showed delayed motor development, calcaneovalgus foot deformities, lower extremity weakness, and mild cognitive delay. At age 3.5 years, she could not run and had an unsteady gait. There was no sensory loss. EMG was consistent with nonlength-dependent motor neuron disease. A sister, who was not studied, reportedly had similar motor delay diagnosed as cerebral palsy, abnormal gait, and polymicrogyria on brain imaging.


.0007 CORTICAL DYSPLASIA, COMPLEX, WITH OTHER BRAIN MALFORMATIONS 13

DYNC1H1, LYS3336ASN
  
RCV000049270...

In a 12-year-old patient (P122) with complex cortical dysplasia with other brain malformations-13 (CDCBM13; 614563), Poirier et al. (2013) identified a de novo heterozygous c.10008G-T transversion in the DYNC1H1 gene, resulting in a lys3336-to-asn (K3336N) substitution at a conserved residue in the microtubule-binding domain. In vitro functional expression studies showed that the mutant protein had decreased microtubule binding affinity compared to wildtype. The mutation was found by whole-exome sequencing and was not present in several large control databases. The patient had microcephaly (-4 SD), early-onset epilepsy, foot deformities consistent with an axonal neuropathy, and was bedridden with spastic tetraplegia. Brain MRI showed posterior pachygyria, frontal polymicrogyria, nodular heterotopia, dysmorphic basal ganglia, and hypoplasia of the corpus callosum, brainstem, and cerebellum.


.0008 CORTICAL DYSPLASIA, COMPLEX, WITH OTHER BRAIN MALFORMATIONS 13

DYNC1H1, ARG3384GLN
  
RCV000049271...

In a 10-year-old patient (P217) with complex cortical dysplasia with other brain malformations-13 (CDCBM13; 614563), Poirier et al. (2013) identified a de novo heterozygous c.10151G-A transition (c.10151G-A, NM_001376) in the DYNC1H1 gene, resulting in an arg3384-to-gln (R3384Q) substitution at a conserved residue in the microtubule-binding domain. In vitro functional expression studies showed that the mutant protein had decreased microtubule binding affinity compared to wildtype. The patient had microcephaly (-4 SD), early-onset epilepsy, foot deformities consistent with an axonal neuropathy, and was bedridden with spastic tetraplegia. Brain MRI showed posterior pachygyria, frontal polymicrogyria, dysmorphic basal ganglia and corpus callosum, and hypoplasia of the brainstem and cerebellum.


.0009 CORTICAL DYSPLASIA, COMPLEX, WITH OTHER BRAIN MALFORMATIONS 13

DYNC1H1, ARG3344GLN
  
RCV000049272...

In 2 unrelated children (P535 and 574C) with complex cortical dysplasia with other brain malformations-13 (CDCBM13; 614563), Poirier et al. (2013) identified a de novo heterozygous c.10031G-A transition (c.10031G-A, NM_001376) in the DYNC1H1 gene, resulting in an arg3344-to-gln (R3344Q) substitution at a conserved residue in the microtubule-binding domain. One patient was a 5-year-old with severe intellectual disability and autistic features, early-onset epileptic encephalopathy, and MRI findings of posterior agyria, nodular heterotopia, and dysmorphic basal ganglia and corpus callosum. The other patient was a 3-year-old with moderate intellectual disability, focal seizures, and MRI findings of posterior pachygyria and small cerebellar vermis.


REFERENCES

  1. Braunstein, K. E., Eschbach, J., Rona-Voros, K., Soylu, R., Mikrouli, E., Larmet, Y., Rene, F., Gonzalez De Aguilar, J.-L., Loeffler, J.-P., Muller, H.-P., Bucher, S., Kaulisch, T., and 10 others. A point mutation in the dynein heavy chain gene leads to striatal atrophy and compromises neurite outgrowth of striatal neurons. Hum. Molec. Genet. 19: 4385-4398, 2010. [PubMed: 20807776, related citations] [Full Text]

  2. Carter, A. P., Garbarino, J. E., Wilson-Kubalek, E. M., Shipley, W. E., Cho, C., Milligan, R. A., Vale, R. D., Gibbons, I. R. Structure and functional role of dynein's microtubule-binding domain. Science 322: 1691-1695, 2008. [PubMed: 19074350, related citations] [Full Text]

  3. Criswell, P. S., Ostrowski, L. E., Asai, D. J. A novel cytoplasmic dynein heavy chain: expression of DHC1b in mammalian ciliated epithelial cells. J. Cell Sci. 109: 1891-1898, 1996. [PubMed: 8832411, related citations] [Full Text]

  4. Dixit, R., Ross, J. L., Goldman, Y. E., Holzbaur, E. L. F. Differential regulation of dynein and kinesin motor proteins by tau. Science 319: 1086-1089, 2008. [PubMed: 18202255, images, related citations] [Full Text]

  5. Eschbach, J., Sinniger, J., Bouitbir, J., Fergani, A., Schlagowski, A.-I., Zoll, J., Geny, B., Rene, F., Larmet, Y., Marion, V., Baloh, R. H., Harms, M. B., Shy, M. E., Messadeq, N., Weydt, P., Loeffler, J.-P., Ludolph, A. C., Dupuis, L. Dynein mutations associated with hereditary motor neuropathies impair mitochondrial morphology and function with age. Neurobiol. Dis. 58: 220-230, 2013. [PubMed: 23742762, images, related citations] [Full Text]

  6. Gibbons, B. H., Asai, D. J., Tang, W.-J. Y., Hays, T. S., Gibbons, I. R. Phylogeny and expression of axonemal and cytoplasmic dynein genes in sea urchins. Molec. Biol. Cell 5: 57-70, 1994. [PubMed: 8186465, related citations] [Full Text]

  7. Hafezparast, M., Klocke, R., Ruhrberg, C., Marquardt, A., Ahmad-Annuar, A., Bowen, S., Lalli, G., Witherden, A. S., Hummerich, H., Nicholson, S., Morgan, P. J., Oozageer, R., and 27 others. Mutations in dynein link motor neuron degeneration to defects in retrograde transport. Science 300: 808-812, 2003. [PubMed: 12730604, related citations] [Full Text]

  8. Harada, A., Takei, Y., Kanai, Y., Tanaka, Y., Nonaka, S., Hirokawa, N. Golgi vesiculation and lysosome dispersion in cells lacking cytoplasmic dynein. J. Cell Biol. 141: 51-59, 1998. [PubMed: 9531547, images, related citations] [Full Text]

  9. Harms, M. B., Allred, P., Gardner, R., Jr., Fernandes Filho, J. A., Florence, J., Pestronk, A., Al-Lozi, M., Baloh, R. H. Dominant spinal muscular atrophy with lower extremity predominance: linkage to 14q32. Neurology 75: 539-546, 2010. [PubMed: 20697106, images, related citations] [Full Text]

  10. Harms, M. B., Ori-McKenney, K. M., Scoto, M., Tuck, E. P., Bell, S., Ma, D., Masi, S., Allred, P., Al-Lozi, M., Reilly, M. M., Miller, L. J., Jani-Acsadi, A., Pestronk, A., Shy, M. E., Muntoni, F., Vallee, R. B., Baloh, R. H. Mutations in the tail domain of DYNC1H1 cause dominant spinal muscular atrophy. Neurology 78: 1714-1720, 2012. [PubMed: 22459677, images, related citations] [Full Text]

  11. Jamuar, S. S., Lam, A. N., Kircher, M., D'Gama, A. M., Wang, J., Barry, B. J., Zhang, X., Hill, R. S., Partlow, J. N., Rozzo, A., Servattalab, S., Mehta, B. K., and 20 others. Somatic mutations in cerebral cortical malformations. New Eng. J. Med. 371: 733-743, 2014. [PubMed: 25140959, images, related citations] [Full Text]

  12. Kural, C., Kim, H., Syed, S., Goshima, G., Gelfand, V. I., Selvin, P. R. Kinesin and dynein move a peroxisome in vivo: a tug-of-war or coordinated movement? Science 308: 1469-1472, 2005. [PubMed: 15817813, related citations] [Full Text]

  13. Mikami, A., Paschal, B. M., Mazumdar, M., Vallee, R. B. Molecular cloning of retrograde transport motor cytoplasmic dynein (MAP 1C). Neuron 10: 787-796, 1993. [PubMed: 7684232, related citations] [Full Text]

  14. Narayan, D., Desai, T., Banks, A., Patanjali, S. R., Ravikumar, T. S., Ward, D. C. Localization of the human cytoplasmic dynein heavy chain (DNECL) to 14qter by fluorescence in situ hybridization. Genomics 22: 660-661, 1994. Note: Erratum: Genomics 24: 618 only, 1994. [PubMed: 8001984, related citations] [Full Text]

  15. Ori-McKenney, K. M., Vallee, R. B. Neuronal migration defects in the Loa dynein mutant mouse. Neural Dev. 6: 26, 2011. Note: Electronic Article. [PubMed: 21612657, images, related citations] [Full Text]

  16. Ori-McKenney, K. M., Xu, J., Gross, S. P., Vallee, R. B. A cytoplasmic dynein tail mutation impairs motor processivity. Nature Cell Biol. 12: 1228-1234, 2010. [PubMed: 21102439, images, related citations] [Full Text]

  17. Pazour, G. J., Agrin, N., Walker, B. L., Witman, G. B. Identification of predicted human outer dynein arm genes: candidates for primary ciliary dyskinesia genes. (Letter) J. Med. Genet. 43: 62-73, 2006. [PubMed: 15937072, images, related citations] [Full Text]

  18. Poirier, K., Lebrun, N., Broix, L., Tian, G., Saillour, Y., Boscheron, C., Parrini, E., Valence, S., Saint Pierre, B., Oger, M., Lacombe, D., Genevieve, D., and 23 others. Mutations in TUBG1, DYNC1H1, KIF5C and KIF2A cause malformations of cortical development and microcephaly. Nature Genet. 45: 639-647, 2013. Note: Erratum: Nature Genet. 45: 962 only, 2013. [PubMed: 23603762, images, related citations] [Full Text]

  19. Sasaki, S., Shionoya, A., Ishida, M., Gambello, M. J., Yingling, J., Wynshaw-Boris, A., Hirotsune, S. A LIS1/NUDEL/cytoplasmic dynein heavy chain complex in the developing and adult nervous system. Neuron 28: 681-696, 2000. [PubMed: 11163259, related citations] [Full Text]

  20. Tsurusaki, Y., Saitoh, S., Tomizawa, K., Sudo, A., Asahina, N., Shiraishi, H., Ito, J., Tanaka, H., Doi, H., Saitsu, H., Miyake, N., Matsumoto, N. A DYNC1H1 mutation causes a dominant spinal muscular atrophy with lower extremity predominance. Neurogenetics 13: 327-332, 2012. [PubMed: 22847149, related citations] [Full Text]

  21. Urnavicius, L., Lau, C. K., Elshenawy, M. M., Morales-Rios, E., Motz, C., Yildiz, A., Carter, A. P. Cryo-EM shows how dynactin recruits two dyneins for faster movement. Nature 554: 202-206, 2018. [PubMed: 29420470, images, related citations] [Full Text]

  22. Vaisberg, E. A., Grissom, P. M., McIntosh, J. R. Mammalian cells express three distinct dynein heavy chains that are localized to different cytoplasmic organelles. J. Cell Biol. 133: 831-842, 1996. [PubMed: 8666668, related citations] [Full Text]

  23. Vaisberg, E. A., Koonce, M. P., McIntosh, J. R. Cytoplasmic dynein plays a role in mammalian mitotic spindle formation. J. Cell Biol. 123: 849-858, 1993. [PubMed: 8227145, related citations] [Full Text]

  24. Vissers, L. E. L. M., de Ligt, J., Gilissen, C., Janssen, I., Steehouwer, M., de Vries, P., van Lier, B., Arts, P., Wieskamp, N., del Rosario, M., van Bon, B. W. M., Hoischen, A., de Vries, B. B. A., Brunner, H. G., Veltman, J. A. A de novo paradigm for mental retardation. Nature Genet. 42: 1109-1112, 2010. [PubMed: 21076407, related citations] [Full Text]

  25. Weedon, M. N., Hastings, R., Caswell, R., Xie, W., Paszkiewicz, K., Antoniadi, T., Williams, M., King, C., Greenhalgh, L., Newbury-Ecob, R., Ellard, S. Exome sequencing identifies a DYNC1H1 mutation in a large pedigree with dominant axonal Charcot-Marie-Tooth disease. Am. J. Hum. Genet. 89: 308-312, 2011. [PubMed: 21820100, images, related citations] [Full Text]

  26. Willemsen, M. H., Vissers, L. E. L., Willemsen, M. A. A. P., van Bon, B. W. M., Kroes, T., de Ligt, J., de Vries, B. B., Schoots, J., Lugtenberg, D., Hamel, B. C. J., van Bokhoven, H., Brunner, H. G., Veltman, J. A., Kleefstra, T. Mutations in DYNC1H1 cause severe intellectual disability with neuronal migration defects. J. Med. Genet. 49: 179-183, 2012. [PubMed: 22368300, related citations] [Full Text]


Ada Hamosh - updated : 04/16/2018
George E. Tiller - updated : 09/12/2017
Ada Hamosh - updated : 09/02/2014
Cassandra L. Kniffin - updated : 7/1/2014
Cassandra L. Kniffin - updated : 9/23/2013
Cassandra L. Kniffin - updated : 6/27/2013
Cassandra L. Kniffin - updated : 4/25/2012
Cassandra L. Kniffin - updated : 4/9/2012
Cassandra L. Kniffin - updated : 9/15/2011
Ada Hamosh - updated : 12/29/2008
Ada Hamosh - updated : 4/4/2008
Patricia A. Hartz - updated : 4/18/2006
Ada Hamosh - updated : 8/2/2005
Ada Hamosh - updated : 5/6/2003
Rebekah S. Rasooly - updated : 11/18/1998
Creation Date:
Victor A. McKusick : 9/13/1994
alopez : 04/07/2023
ckniffin : 04/07/2023
alopez : 04/01/2022
alopez : 04/01/2022
alopez : 04/16/2018
carol : 01/05/2018
alopez : 09/12/2017
alopez : 09/02/2014
carol : 7/2/2014
mcolton : 7/2/2014
ckniffin : 7/1/2014
carol : 9/26/2013
tpirozzi : 9/26/2013
ckniffin : 9/23/2013
carol : 8/28/2013
carol : 7/10/2013
carol : 7/9/2013
ckniffin : 6/27/2013
carol : 9/19/2012
terry : 7/5/2012
terry : 5/2/2012
carol : 4/27/2012
ckniffin : 4/25/2012
alopez : 4/10/2012
terry : 4/10/2012
terry : 4/10/2012
ckniffin : 4/9/2012
carol : 9/16/2011
ckniffin : 9/15/2011
alopez : 12/29/2008
terry : 12/29/2008
alopez : 4/11/2008
terry : 4/4/2008
mgross : 4/19/2006
mgross : 4/18/2006
alopez : 8/3/2005
terry : 8/2/2005
carol : 5/12/2004
alopez : 5/8/2003
terry : 5/6/2003
dkim : 12/18/1998
dkim : 12/3/1998
psherman : 12/2/1998
alopez : 11/18/1998
alopez : 8/21/1998
mimadm : 9/23/1995
carol : 9/13/1994

* 600112

DYNEIN, CYTOPLASMIC 1, HEAVY CHAIN 1; DYNC1H1


Alternative titles; symbols

DNCH1
DYNEIN, CYTOPLASMIC-LIKE; DNCL; DNECL
DYNEIN HEAVY POLYPEPTIDE, CYTOPLASMIC; DHC1; DHC1A
DNCHC1


HGNC Approved Gene Symbol: DYNC1H1

SNOMEDCT: 782829002;  


Cytogenetic location: 14q32.31     Genomic coordinates (GRCh38): 14:101,964,573-102,056,443 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
14q32.31 Charcot-Marie-Tooth disease, axonal, type 2O 614228 Autosomal dominant 3
Cortical dysplasia, complex, with other brain malformations 13 614563 Autosomal dominant 3
Spinal muscular atrophy, lower extremity-predominant 1, AD 158600 Autosomal dominant 3

TEXT

Description

The DYNC1H1 gene encodes a large (over 530 kD) crucial subunit of the cytoplasmic dynein complex (summary by Poirier et al., 2013). Dyneins are a group of microtubule-activated ATPases that serve to convert chemical energy into mechanical energy. They have been divided into 2 large subgroups, namely, the axonemal and cytoplasmic dyneins. Cytoplasmic dynein has been implicated in a variety of other forms of intracellular motility, including retrograde axonal transport, protein sorting between apical and basolateral surfaces, and redistribution of organelles like endosomes and lysosomes. Molecules of conventional cytoplasmic dynein contain 2 heavy chain polypeptides and a number of intermediate and light chains. They sediment at approximately 20S (Vaisberg et al., 1993).


Cloning and Expression

Mikami et al. (1993) isolated cDNAs encoding the heavy chain of MAP1C, a rat cytoplasmic dynein. The predicted 4,644-amino acid protein contains 4 ATP-binding consensus sequences. Southern blot analysis suggested that there is a single cytoplasmic dynein gene in rat. Gibbons et al. (1994) identified DYH1a, a sea urchin cytoplasmic dynein with homology to MAP1C. Criswell et al. (1996) reported that MAP1C, or DHC1a, expression was unchanged during ciliogenesis in primary rat tracheal epithelial cells.

By screening an adenocarcinoma cell line library with p22, Vaisberg et al. (1996) isolated a DHC1 cDNA. The predicted partial protein sequence shares 99% and 34% identity with rat DHC1a and human DNHC2 (603297), respectively. Antibodies against DHC1 recognized a high molecular mass protein on Western blots of extracts from several mammalian cell lines. Northern blot analysis revealed that DHC1 is expressed as an approximately 15-kb mRNA in several mammalian cells lines and human tissues, including those that make neither cilia nor flagella. By immunofluorescence, Vaisberg et al. (1996) found that DHC1 localizes in a punctate pattern in the cytoplasm that is generally brighter in the perinuclear area and dimmer near the cell periphery. DHC1 redistributes during mitosis to the kinetochores and mitotic spindle.


Gene Function

Vaisberg et al. (1993) cloned a partial cDNA (p22) encoding the putative ATP-hydrolytic site of the human conventional cytoplasmic dynein heavy chain (DHC). Antibodies against the resulting polypeptide inhibited dynein motor activity in vitro. Injection of these antibodies into mitotic mammalian cells blocked the formation of spindles in prophase or during recovery from nocodazole treatment at later stages of mitosis. The cells became arrested with unseparated centrosomes and formed monopolar spindles. However, there was no detectable effect on chromosome attachment to a bipolar spindle or on chromosome movements during anaphase. Vaisberg et al. (1993) suggested that cytoplasmic dynein plays a unique and important role in the initial events of bipolar spindle formation, while any later roles in mitosis may be redundant.

Sasaki et al. (2000) demonstrated that Lis1 (PAFAH1B1; 601545) directly interacts with Dync1h1 and Ndel1 (607538) in the developing mouse brain. Lis1 and Ndel1 colocalized predominantly at the centrosome in early neuroblasts, but redistributed to axons in association with retrograde dynein motor proteins. Ndel1 and Lis1 regulated the distribution of Dync1h1 along microtubules, establishing a direct functional link between LIS1, NDEL1, and microtubule motors during neuronal migration and axonal retrograde transport in the mammalian brain.

Kural et al. (2005) used fluorescence imaging with 1-nanometer accuracy (FIONA) to analyze organelle movement by conventional kinesin (602809) and cytoplasmic dynein in a cell. They located a green fluorescent protein (GFP)-tagged peroxisome in cultured Drosophila S2 cells to within 1.5 nanometers in 1.1 milliseconds, a 400-fold improvement in temporal resolution, sufficient to determine the average step size to be approximately 8 nanometers for both dynein and kinesin. Furthermore, Kural et al. (2005) found that dynein and kinesin do not work against each other in vivo during peroxisome transport. Rather, multiple kinesins or multiple dyneins work together, producing up to 10 times the in vitro speed.

To determine the effects of tau (157140) on dynein and kinesin motility, Dixit et al. (2008) conducted single-molecule studies of motor proteins moving along tau-decorated microtubules. Dynein tended to reverse direction, whereas kinesin tended to detach at patches of bound tau. Kinesin was inhibited at about a tenth of the tau concentration that inhibited dynein, and the microtubule-binding domain of tau was sufficient to inhibit motor activity. The differential modulation of dynein and kinesin motility suggested that microtubule-associated proteins (MAPs) can spatially regulate the balance of microtubule-dependent axonal transport.


Mapping

By fluorescence in situ hybridization, Narayan et al. (1994) localized the heavy chain gene of human cytoplasmic dynein to the terminal region of 14q. Indirect confirmation of this result was provided by the fact that the mouse cytoplasmic dynein heavy chain had been localized to murine chromosome 12, which shows extensive homology of synteny with human 14 (Mikami et al., 1993).

By genomic sequence analysis, Pazour et al. (2006) mapped the DNCH1 gene to chromosome 14q32.


Biochemical Features

Crystal Structure

Carter et al. (2008) reported the crystal structure of the mouse cytoplasmic dynein microtubule binding domain (MTBD) and a portion of the coiled coil, which supports a mechanism by which the ATPase domain and the MTBD may communicate through a shift in the heptad registry of the coiled coil. Surprisingly, functional data suggested that the MTBD and not the ATPase domain is the main determinant of the direction of dynein motility.

Urnavicius et al. (2018) used electron microscopy and single-molecule studies to show that adaptors can recruit a second dynein to dynactin (601143). Whereas BICD2 (609797) is biased towards recruiting a single dynein, the adaptors BICDR1 (617002) and HOOK3 (607825) predominantly recruit 2 dyneins. Urnavicius et al. (2018) found that the shift towards a double dynein complex increases both the force and speed of the microtubule motor. The 3.5-angstrom resolution cryoelectron microscopy reconstruction of a dynein tail-dynactin-BICDR1 complex revealed how dynactin can act as a scaffold to coordinate 2 dyneins side by side. Urnavicius et al. (2018) concluded that their work provided a structural basis for understanding how diverse adaptors recruit different numbers of dyneins and regulate the motile properties of the dynein-dynactin transport machine.


Molecular Genetics

Charcot-Marie-Tooth Disease, Axonal, Type 2O

In affected members of a large 4-generation family with autosomal dominant axonal Charcot-Marie-Tooth disease type 2O (CMT2O; 614228), Weedon et al. (2011) identified a heterozygous mutation in the DYNC1H1 gene (H306R; 600112.0001). The mutation was identified by exome sequencing. Affected individuals had onset in childhood of delayed motor milestones and abnormal gait and falls associated with distal lower limb weakness and wasting and distal sensory impairment. Weedon et al. (2011) noted that mouse models had implicated mutations in this gene in neuropathic disease.

Complex Cortical Dysplasia With Other Brain Malformations 13

By family-based exome sequencing of 10 case-parent trios with global developmental delay, Vissers et al. (2010) identified a de novo heterozygous mutation in the DYNC1H1 gene (H3822P; 600112.0002) in 1 patient. He had hypotonia and mild dysmorphic facial features. Follow-up of the patient at age 6 years by Willemsen et al. (2012) noted that he had hypotonia, hyporeflexia, and broad-based waddling gait with toe-walking. Reevaluation of brain MRI showed complex cortical dysplasia with other brain malformations (CDCBM13; 614563). Willemsen et al. (2012) identified a second de novo heterozygous mutation in the DYNC1H1 gene (E1518K; 600112.0003) in a 51-year-old woman with severe developmental delay since infancy and an inability to walk or speak. She had mild dysmorphic features, seizures, and spastic tetraplegia. Cerebral CT scan at the age 46 years showed enlarged ventricles and clear signs of cortical malformation with wide opercular regions and an abnormal flat cortex with only a few simple and shallow sulci; MRI scan was not possible. Willemsen et al. (2012) noted that DYNC1H1 interacts with LIS1 (601545), haploinsufficiency of which results in the severe neuronal migration disorder lissencephaly-1 (607432), and that Dync1h1 mutant mice show neuronal migration defects (Ori-McKenney and Vallee, 2011), providing evidence of the pathogenicity of the mutations. Willemsen et al. (2012) also noted that their 2 patients showed variable signs consistent with peripheral neuropathy and that some of the patients with CMT2O (Weedon et al., 2011) showed learning difficulties, indicating that DYNC1H1 mutations may result in a broad neurologic phenotypic spectrum.

Poirier et al. (2013) identified 8 different de novo heterozygous mutations in the DYNC1H1 gene (see, e.g., 600112.0007-600112.0009) in 8 unrelated patients ascertained for evaluation due to malformations of cortical development (CDCBM13). The patients had global developmental delay, and most had early-onset seizures. Mutations in the first several patients were found by whole-exome sequencing, whereas subsequent patients were identified by direct sequencing of this gene in a larger cohort of affected individuals. In vitro functional expression studies of 2 of the variants showed that the mutant proteins had decreased microtubule binding affinity compared to wildtype. In addition, there was 1 family in which a mother and her 2 children carried a missense variant (K3241T): 1 of the children had mild intellectual disability, but the mother and the other child had normal cognition. All 3 were normocephalic, showed posterior pachygyria, and had focal seizures. No functional studies were performed on the K3241T variant, which occurred at a nonconserved residue.

Jamuar et al. (2014) used a customized panel of known and candidate genes associated with brain malformations to apply targeted high-coverage sequencing (depth greater than or equal to 200x) to leukocyte-derived DNA samples from 158 individuals with brain malformations. They found potentially causal mutations in the candidate gene DYNC1H1 in 2 individuals with pachygyria; in a parallel study they found de novo mutations in DYNC1H1 in 2 other individuals with pachygyria. The 4 individuals had strikingly similar MRI findings, with posterior-predominant pachygyria, thickened cortex in the perisylvian region, and mildly dysmorphic corpus callosum (CDCBM13).

Lower Extremity-Predominant Spinal Muscular Atrophy 1, Autosomal Dominant

In affected members of a large family with autosomal dominant lower extremity-predominant spinal muscular atrophy-1 (SMALED1; 158600) originally reported by Harms et al. (2010), Harms et al. (2012) identified a heterozygous mutation in the DYNC1H1 gene (I584L; 600112.0004). Patient skin fibroblasts showed normal binding to microtubules in the absence of ATP, but markedly decreased binding to microtubules in the presence of ATP. The mutant dynein also appeared to disrupt the stability of the dynein complex. Two additional families with a similar disorder were found to carry heterozygous DYNC1H1 mutations (600112.0005 and 600112.0006). The findings were similar to those observed in Loa homozygous mice (Hafezparast et al., 2003).

In 2 Japanese sibs with autosomal dominant lower extremity spinal muscular atrophy and no sensory symptoms, Tsurusaki et al. (2012) identified a heterozygous missense mutation in the DYNC1H1 gene (H306R; 600112.0001). The mutation, which was found by exome sequencing, was inherited from their mother, who had mild symptoms. The same mutation had previously been found by Weedon et al. (2011) in a family with autosomal dominant axonal Charcot-Marie-Tooth disease type 2O (CMT2O; 614228).

Based on findings in mutant mice, Eschbach et al. (2013) studied the mitochondria in fibroblasts derived from SMALED1 patients with the K671E (600112.0005) and I584L mutations. Both cell lines showed intensely fragmented mitochondria, with smaller mitochondria in K671E cells and increased areas of individual mitochondria in I584L cells. Both cell lines also showed decreased levels of mitofusin-1 (MFN1; 608506). Eschbach et al. (2013) suggested that dysfunction of mitochondrial transport may contribute to disease pathogenesis in patients with DYNC1H1 mutations.


Animal Model

Harada et al. (1998) generated Dnchc1 knockout mice by targeted disruption. No embryos were identified at 8.5 days postcoitum. Cultured blastocysts of Dnchc1-null embryos demonstrated a highly vesiculated Golgi complex that was distributed throughout the cytoplasm. Endosomes and lysosomes were not concentrated near the nucleus but were distributed evenly throughout the cytoplasm.

'Legs at odd angles' (Loa) and 'Cramping 1' (cra1) are mouse phenotypes that arose from ENU (N-ethyl-N-nitrosourea) mutagenesis. They are transmitted as autosomal dominant traits and give rise to age-related progressive loss of muscle tone and locomotor ability in heterozygous mice without a major reduction in life span. Homozygous mice show a more severe phenotype with an inability to feed and move, and die within 24 hours of birth. Hafezparast et al. (2003) showed that Loa is caused by a T-to-A transversion in the Dnchc1 gene that changes the phenylalanine at codon 580 to a tyrosine. The Cra1 phenotype results from an A-to-G transition leading to a tyrosine-to-cysteine substitution at codon 1055 of the Dnchc1 gene. Intercrossing heterozygous Cra1/+ with Loa/+ heterozygous mice yielded compound heterozygotes that died shortly after birth, demonstrating that the phenotypes are allelic. Homozygous and compound heterozygous mice developed Lewy-like inclusion bodies, and homozygous mice displayed significant loss of spinal anterior horn cells in utero. The Loa and Cra1 mutations do not cause overt deficits across the range of known DNCHC1 functions but result in a specific defect in fast retrograde transport that appears to manifest only in alpha motor neurons. Hafezparast et al. (2003) concluded that the Loa and Cra1 mutations exhibit remarkable similarities to specific features of human pathology for amyotrophic lateral sclerosis (ALS; 105400) and other motor neuron degeneration phenotypes including Lewy body-like inclusions containing SOD1 (147450), CDK5 (123831), neurofilaments, and ubiquitin (191339).

Braunstein et al. (2010) studied the 'cramping' mouse (Cra), which is heterozygous for a tyr1055-to-cys mutation in Dync1h1 that impairs retrograde axonal transport. These mice exhibited motor and behavioral abnormalities including hindlimb clasping, early muscle weakness, incoordination, and hyperactivity. In vivo brain imaging using magnetic resonance imaging showed striatal atrophy and lateral ventricle enlargement. In the striatum, altered dopamine signaling, decreased dopamine D1 receptor (DRD1; 126449) and D2 receptor (DRD2; 126450) binding in positron emission tomography SCAN, and prominent astrocytosis were observed, although there was no neuronal loss either in the striatum or substantia nigra. In vitro, dynein mutant striatal neurons displayed strongly impaired neuritic morphology. The authors concluded that dynein is required for the normal morphology and function of striatal neurons, and may play a role in the pathogenesis of neurodegenerative disorders of the basal ganglia such as Perry syndrome (168605) and Huntington disease (HD; 143100).

Eschbach et al. (2013) found that cultured embryonic fibroblasts from Cra1 mice had profoundly disrupted mitochondrial networks, including fragmented mitochondria and mitochondrial aggregates, associated with decreased mitofusin-1 (MFN1; 608506). Skeletal muscle from these mice showed progressive mitochondrial dysfunction with decreased respiration and altered energy metabolism. In addition, mutant mice developed late-onset glucose intolerance consistent with mitochondrial dysfunction. Dync1h1 mutant fibroblasts showed impaired perinuclear clustering of mitochondria in response to mitochondrial uncoupling. These findings indicated that dynein function is required for the maintenance of mitochondrial morphology and function.

Ori-McKenney and Vallee (2011) demonstrated that Loa homozygous mice had defects in neocortical lamination and neuronal migration resulting from a reduction in the rate of radial migration of bipolar neurons. Examination of brains from mutant mice showed blurred laminar boundaries, indicating cortical disorganization. The hippocampal dentate gyrus was smaller than wildtype. There was also a decrease in axonal extension within the brain, indicating that dynein processivity is necessary for axon elongation. The findings were similar, but less severe, than those reported in Lis1 (601545) compound heterozygous mutant mice.


ALLELIC VARIANTS 9 Selected Examples):

.0001   CHARCOT-MARIE-TOOTH DISEASE, AXONAL, TYPE 2O

SPINAL MUSCULAR ATROPHY, LOWER EXTREMITY-PREDOMINANT, 1, AUTOSOMAL DOMINANT, INCLUDED
DYNC1H1, HIS306ARG
SNP: rs387906738, ClinVar: RCV000022929, RCV000055662, RCV000192255, RCV001091151

Charcot-Marie-Tooth Disease, Axonal, Type 2O

In affected members of a large 4-generation family with autosomal dominant axonal Charcot-Marie-Tooth disease type 2O (CMT2O; 614228), Weedon et al. (2011) identified a heterozygous 917A-G transition in the DYNC1H1 gene, resulting in a his306-to-arg (H306R) substitution at a highly conserved residue in the homodimerization domain. Affected individuals had onset in childhood of delayed motor milestones and abnormal gait and falls associated with distal lower limb weakness and wasting and distal sensory impairment. Weedon et al. (2011) noted that mouse models had implicated mutations in this gene in neuropathic disease.

Spinal Muscular Atrophy, Lower Extremity-Predominant 1, Autosomal Dominant

Tsurusaki et al. (2012) identified a heterozygous H306R mutation in the DYNC1H1 gene in 2 Japanese sibs with autosomal dominant lower extremity-predominant spinal muscular atrophy-1 (SMALED1; 158600) without sensory impairment. The mother, who had mild symptoms, also carried the mutation. The mutation, which was found by exome sequencing and confirmed by Sanger sequencing, was not present in the dbSNP or 1000 Genomes databases, in 33 in-house exomes, or in 177 Japanese control individuals.


.0002   CORTICAL DYSPLASIA, COMPLEX, WITH OTHER BRAIN MALFORMATIONS 13

DYNC1H1, HIS3822PRO
SNP: rs387906739, ClinVar: RCV000022930, RCV003447090

In a 4-year-old boy with complex cortical dysplasia with other brain malformations-13 (CDCBM13; 614563), Vissers et al. (2010) identified a de novo heterozygous c.11465A-C transversion (c.11465A-C, NM_001376) in the DYNC1H1 gene, resulting in a his3822-to-pro (H3822P) substitution at a highly conserved residue in the stem domain of the protein. The mutation was found by family-based exome sequencing, and was not found in 1,664 control chromosomes. The patient showed hypotonia at age 6 months, followed by delayed psychomotor development. Mild dysmorphic features included prominent forehead, plagiocephaly, hypotonic face with downslanting palpebral fissures, and short, broad hands and feet. Brain MRI was reported as normal. Follow-up of the patient at age 6 years by Willemsen et al. (2012) noted that he had hypotonia, hyporeflexia, and broad-based waddling gait with toe-walking. Reevaluation of brain MRI showed signs of bilateral cortical malformation with deficient gyration of the frontal lobes and an area suggestive of focal cortical dysplasia, consistent with a neuronal migration defect.


.0003   CORTICAL DYSPLASIA, COMPLEX, WITH OTHER BRAIN MALFORMATIONS 13

DYNC1H1, GLU1518LYS
SNP: rs387906740, ClinVar: RCV000022931, RCV003447091

In a 51-year-old woman with complex cortical dysplasia with other brain malformations-13 (CDCBM13; 614563), Willemsen et al. (2012) identified a de novo heterozygous c.4552G-A transition (c.4552G-A, NM_001376.4) in the DYNC1H1 gene, resulting in a glu1518-to-lys (E1518K) substitution at a highly conserved residue in the motor domain of the protein. The mutation was not found in 445 control exomes. She had severe developmental delay with an inability to walk or speak and generalized seizures since age 3 years. Craniofacial features included brachycephaly, prominent forehead, hypertelorism, deep-set eyes, wide mouth with everted lower lip, and downturned corners of the mouth. Other features included short stature, microcephaly, clubfeet, small hands and feet with short toes, kyphoscoliosis, spastic tetraplegia, and swallowing difficulties. Cerebral CT scan at the age 46 years showed enlarged ventricles and clear signs of cortical malformation with wide opercular regions and an abnormal flat cortex with only a few simple and shallow sulci; MRI scan was not possible.


.0004   SPINAL MUSCULAR ATROPHY, LOWER EXTREMITY-PREDOMINANT, 1, AUTOSOMAL DOMINANT

DYNC1H1, ILE584LEU
SNP: rs387906741, ClinVar: RCV000022932, RCV000789730, RCV003447092

In affected members of a large family with autosomal dominant lower extremity-predominant spinal muscular atrophy (SMALED1; 158600) originally reported by Harms et al. (2010), Harms et al. (2012) identified a heterozygous 1750A-C transversion in exon 8 of the DYNC1H1 gene, resulting in an ile584-to-leu (I584L) substitution at a highly conserved residue in the tail domain of the dynein heavy chain, a highly conserved region critical for organizing multiple dynein subunits into a complex. The mutation was not found in 500 controls or the 1000 Genomes Project. Affected individuals had early-childhood onset of proximal leg weakness with muscle atrophy and nonlength-dependent motor neuron disease without sensory involvement. Patient skin fibroblasts showed normal binding to microtubules in the absence of ATP, but markedly decreased binding to microtubules in the presence of ATP. The mutant dynein also appeared to disrupt the stability of the dynein complex. The findings were similar to those observed in Loa homozygous mice (Hafezparast et al., 2003; Ori-McKenney et al., 2010).


.0005   SPINAL MUSCULAR ATROPHY, LOWER EXTREMITY-PREDOMINANT, 1, AUTOSOMAL DOMINANT

DYNC1H1, LYS671GLU
SNP: rs387906742, ClinVar: RCV000022933, RCV003447093

In affected members of a 3-generation family with autosomal dominant lower extremity-predominant spinal muscular atrophy (SMALED1; 158600), Harms et al. (2012) identified a heterozygous 2011A-G transition in exon 8 of the DYNC1H1 gene, resulting in a lys671-to-glu (K671E) substitution at a highly conserved residue in the tail domain of the dynein heavy chain. The mutation was not found in 500 controls or the 1000 Genomes Project. The 3 affected individuals showed waddling gait from early childhood, with awkward running due to lower limb weakness; upper limbs were not affected. Muscle atrophy and weakness confined to the lower limbs showed little progression throughout life. There was a notable strength discrepancy between knee extension and flexion, with the quadriceps showing significant weakness. Deep tendon reflexes were reduced at the knees, but normal elsewhere. Nerve conduction studies showed small motor responses and normal sensory responses; EMG showed chronic denervation. One patient had heel cord contractures and inturning feet, whereas another had fasciculations of the calves.


.0006   SPINAL MUSCULAR ATROPHY, LOWER EXTREMITY-PREDOMINANT, 1, AUTOSOMAL DOMINANT

DYNC1H1, TYR970CYS
SNP: rs387906743, ClinVar: RCV000022934, RCV003447094

In a 3.5-year-old girl with autosomal dominant lower extremity-predominant spinal muscular atrophy (SMALED1; 158600), Harms et al. (2012) identified a heterozygous 3170A-G transition in exon 11 of the DYNC1H1 gene, resulting in a tyr970-to-cys (Y970C) substitution at a highly conserved residue. The mutation was not found in 500 controls or the 1000 Genomes Project. The patient showed delayed motor development, calcaneovalgus foot deformities, lower extremity weakness, and mild cognitive delay. At age 3.5 years, she could not run and had an unsteady gait. There was no sensory loss. EMG was consistent with nonlength-dependent motor neuron disease. A sister, who was not studied, reportedly had similar motor delay diagnosed as cerebral palsy, abnormal gait, and polymicrogyria on brain imaging.


.0007   CORTICAL DYSPLASIA, COMPLEX, WITH OTHER BRAIN MALFORMATIONS 13

DYNC1H1, LYS3336ASN
SNP: rs397509410, ClinVar: RCV000049270, RCV003447108

In a 12-year-old patient (P122) with complex cortical dysplasia with other brain malformations-13 (CDCBM13; 614563), Poirier et al. (2013) identified a de novo heterozygous c.10008G-T transversion in the DYNC1H1 gene, resulting in a lys3336-to-asn (K3336N) substitution at a conserved residue in the microtubule-binding domain. In vitro functional expression studies showed that the mutant protein had decreased microtubule binding affinity compared to wildtype. The mutation was found by whole-exome sequencing and was not present in several large control databases. The patient had microcephaly (-4 SD), early-onset epilepsy, foot deformities consistent with an axonal neuropathy, and was bedridden with spastic tetraplegia. Brain MRI showed posterior pachygyria, frontal polymicrogyria, nodular heterotopia, dysmorphic basal ganglia, and hypoplasia of the corpus callosum, brainstem, and cerebellum.


.0008   CORTICAL DYSPLASIA, COMPLEX, WITH OTHER BRAIN MALFORMATIONS 13

DYNC1H1, ARG3384GLN
SNP: rs397509411, ClinVar: RCV000049271, RCV001091160, RCV003447109

In a 10-year-old patient (P217) with complex cortical dysplasia with other brain malformations-13 (CDCBM13; 614563), Poirier et al. (2013) identified a de novo heterozygous c.10151G-A transition (c.10151G-A, NM_001376) in the DYNC1H1 gene, resulting in an arg3384-to-gln (R3384Q) substitution at a conserved residue in the microtubule-binding domain. In vitro functional expression studies showed that the mutant protein had decreased microtubule binding affinity compared to wildtype. The patient had microcephaly (-4 SD), early-onset epilepsy, foot deformities consistent with an axonal neuropathy, and was bedridden with spastic tetraplegia. Brain MRI showed posterior pachygyria, frontal polymicrogyria, dysmorphic basal ganglia and corpus callosum, and hypoplasia of the brainstem and cerebellum.


.0009   CORTICAL DYSPLASIA, COMPLEX, WITH OTHER BRAIN MALFORMATIONS 13

DYNC1H1, ARG3344GLN
SNP: rs397509412, ClinVar: RCV000049272, RCV001255327, RCV001262933, RCV001291070

In 2 unrelated children (P535 and 574C) with complex cortical dysplasia with other brain malformations-13 (CDCBM13; 614563), Poirier et al. (2013) identified a de novo heterozygous c.10031G-A transition (c.10031G-A, NM_001376) in the DYNC1H1 gene, resulting in an arg3344-to-gln (R3344Q) substitution at a conserved residue in the microtubule-binding domain. One patient was a 5-year-old with severe intellectual disability and autistic features, early-onset epileptic encephalopathy, and MRI findings of posterior agyria, nodular heterotopia, and dysmorphic basal ganglia and corpus callosum. The other patient was a 3-year-old with moderate intellectual disability, focal seizures, and MRI findings of posterior pachygyria and small cerebellar vermis.


REFERENCES

  1. Braunstein, K. E., Eschbach, J., Rona-Voros, K., Soylu, R., Mikrouli, E., Larmet, Y., Rene, F., Gonzalez De Aguilar, J.-L., Loeffler, J.-P., Muller, H.-P., Bucher, S., Kaulisch, T., and 10 others. A point mutation in the dynein heavy chain gene leads to striatal atrophy and compromises neurite outgrowth of striatal neurons. Hum. Molec. Genet. 19: 4385-4398, 2010. [PubMed: 20807776] [Full Text: https://doi.org/10.1093/hmg/ddq361]

  2. Carter, A. P., Garbarino, J. E., Wilson-Kubalek, E. M., Shipley, W. E., Cho, C., Milligan, R. A., Vale, R. D., Gibbons, I. R. Structure and functional role of dynein's microtubule-binding domain. Science 322: 1691-1695, 2008. [PubMed: 19074350] [Full Text: https://doi.org/10.1126/science.1164424]

  3. Criswell, P. S., Ostrowski, L. E., Asai, D. J. A novel cytoplasmic dynein heavy chain: expression of DHC1b in mammalian ciliated epithelial cells. J. Cell Sci. 109: 1891-1898, 1996. [PubMed: 8832411] [Full Text: https://doi.org/10.1242/jcs.109.7.1891]

  4. Dixit, R., Ross, J. L., Goldman, Y. E., Holzbaur, E. L. F. Differential regulation of dynein and kinesin motor proteins by tau. Science 319: 1086-1089, 2008. [PubMed: 18202255] [Full Text: https://doi.org/10.1126/science.1152993]

  5. Eschbach, J., Sinniger, J., Bouitbir, J., Fergani, A., Schlagowski, A.-I., Zoll, J., Geny, B., Rene, F., Larmet, Y., Marion, V., Baloh, R. H., Harms, M. B., Shy, M. E., Messadeq, N., Weydt, P., Loeffler, J.-P., Ludolph, A. C., Dupuis, L. Dynein mutations associated with hereditary motor neuropathies impair mitochondrial morphology and function with age. Neurobiol. Dis. 58: 220-230, 2013. [PubMed: 23742762] [Full Text: https://doi.org/10.1016/j.nbd.2013.05.015]

  6. Gibbons, B. H., Asai, D. J., Tang, W.-J. Y., Hays, T. S., Gibbons, I. R. Phylogeny and expression of axonemal and cytoplasmic dynein genes in sea urchins. Molec. Biol. Cell 5: 57-70, 1994. [PubMed: 8186465] [Full Text: https://doi.org/10.1091/mbc.5.1.57]

  7. Hafezparast, M., Klocke, R., Ruhrberg, C., Marquardt, A., Ahmad-Annuar, A., Bowen, S., Lalli, G., Witherden, A. S., Hummerich, H., Nicholson, S., Morgan, P. J., Oozageer, R., and 27 others. Mutations in dynein link motor neuron degeneration to defects in retrograde transport. Science 300: 808-812, 2003. [PubMed: 12730604] [Full Text: https://doi.org/10.1126/science.1083129]

  8. Harada, A., Takei, Y., Kanai, Y., Tanaka, Y., Nonaka, S., Hirokawa, N. Golgi vesiculation and lysosome dispersion in cells lacking cytoplasmic dynein. J. Cell Biol. 141: 51-59, 1998. [PubMed: 9531547] [Full Text: https://doi.org/10.1083/jcb.141.1.51]

  9. Harms, M. B., Allred, P., Gardner, R., Jr., Fernandes Filho, J. A., Florence, J., Pestronk, A., Al-Lozi, M., Baloh, R. H. Dominant spinal muscular atrophy with lower extremity predominance: linkage to 14q32. Neurology 75: 539-546, 2010. [PubMed: 20697106] [Full Text: https://doi.org/10.1212/WNL.0b013e3181ec800c]

  10. Harms, M. B., Ori-McKenney, K. M., Scoto, M., Tuck, E. P., Bell, S., Ma, D., Masi, S., Allred, P., Al-Lozi, M., Reilly, M. M., Miller, L. J., Jani-Acsadi, A., Pestronk, A., Shy, M. E., Muntoni, F., Vallee, R. B., Baloh, R. H. Mutations in the tail domain of DYNC1H1 cause dominant spinal muscular atrophy. Neurology 78: 1714-1720, 2012. [PubMed: 22459677] [Full Text: https://doi.org/10.1212/WNL.0b013e3182556c05]

  11. Jamuar, S. S., Lam, A. N., Kircher, M., D'Gama, A. M., Wang, J., Barry, B. J., Zhang, X., Hill, R. S., Partlow, J. N., Rozzo, A., Servattalab, S., Mehta, B. K., and 20 others. Somatic mutations in cerebral cortical malformations. New Eng. J. Med. 371: 733-743, 2014. [PubMed: 25140959] [Full Text: https://doi.org/10.1056/NEJMoa1314432]

  12. Kural, C., Kim, H., Syed, S., Goshima, G., Gelfand, V. I., Selvin, P. R. Kinesin and dynein move a peroxisome in vivo: a tug-of-war or coordinated movement? Science 308: 1469-1472, 2005. [PubMed: 15817813] [Full Text: https://doi.org/10.1126/science.1108408]

  13. Mikami, A., Paschal, B. M., Mazumdar, M., Vallee, R. B. Molecular cloning of retrograde transport motor cytoplasmic dynein (MAP 1C). Neuron 10: 787-796, 1993. [PubMed: 7684232] [Full Text: https://doi.org/10.1016/0896-6273(93)90195-w]

  14. Narayan, D., Desai, T., Banks, A., Patanjali, S. R., Ravikumar, T. S., Ward, D. C. Localization of the human cytoplasmic dynein heavy chain (DNECL) to 14qter by fluorescence in situ hybridization. Genomics 22: 660-661, 1994. Note: Erratum: Genomics 24: 618 only, 1994. [PubMed: 8001984] [Full Text: https://doi.org/10.1006/geno.1994.1447]

  15. Ori-McKenney, K. M., Vallee, R. B. Neuronal migration defects in the Loa dynein mutant mouse. Neural Dev. 6: 26, 2011. Note: Electronic Article. [PubMed: 21612657] [Full Text: https://doi.org/10.1186/1749-8104-6-26]

  16. Ori-McKenney, K. M., Xu, J., Gross, S. P., Vallee, R. B. A cytoplasmic dynein tail mutation impairs motor processivity. Nature Cell Biol. 12: 1228-1234, 2010. [PubMed: 21102439] [Full Text: https://doi.org/10.1038/ncb2127]

  17. Pazour, G. J., Agrin, N., Walker, B. L., Witman, G. B. Identification of predicted human outer dynein arm genes: candidates for primary ciliary dyskinesia genes. (Letter) J. Med. Genet. 43: 62-73, 2006. [PubMed: 15937072] [Full Text: https://doi.org/10.1136/jmg.2005.033001]

  18. Poirier, K., Lebrun, N., Broix, L., Tian, G., Saillour, Y., Boscheron, C., Parrini, E., Valence, S., Saint Pierre, B., Oger, M., Lacombe, D., Genevieve, D., and 23 others. Mutations in TUBG1, DYNC1H1, KIF5C and KIF2A cause malformations of cortical development and microcephaly. Nature Genet. 45: 639-647, 2013. Note: Erratum: Nature Genet. 45: 962 only, 2013. [PubMed: 23603762] [Full Text: https://doi.org/10.1038/ng.2613]

  19. Sasaki, S., Shionoya, A., Ishida, M., Gambello, M. J., Yingling, J., Wynshaw-Boris, A., Hirotsune, S. A LIS1/NUDEL/cytoplasmic dynein heavy chain complex in the developing and adult nervous system. Neuron 28: 681-696, 2000. [PubMed: 11163259] [Full Text: https://doi.org/10.1016/s0896-6273(00)00146-x]

  20. Tsurusaki, Y., Saitoh, S., Tomizawa, K., Sudo, A., Asahina, N., Shiraishi, H., Ito, J., Tanaka, H., Doi, H., Saitsu, H., Miyake, N., Matsumoto, N. A DYNC1H1 mutation causes a dominant spinal muscular atrophy with lower extremity predominance. Neurogenetics 13: 327-332, 2012. [PubMed: 22847149] [Full Text: https://doi.org/10.1007/s10048-012-0337-6]

  21. Urnavicius, L., Lau, C. K., Elshenawy, M. M., Morales-Rios, E., Motz, C., Yildiz, A., Carter, A. P. Cryo-EM shows how dynactin recruits two dyneins for faster movement. Nature 554: 202-206, 2018. [PubMed: 29420470] [Full Text: https://doi.org/10.1038/nature25462]

  22. Vaisberg, E. A., Grissom, P. M., McIntosh, J. R. Mammalian cells express three distinct dynein heavy chains that are localized to different cytoplasmic organelles. J. Cell Biol. 133: 831-842, 1996. [PubMed: 8666668] [Full Text: https://doi.org/10.1083/jcb.133.4.831]

  23. Vaisberg, E. A., Koonce, M. P., McIntosh, J. R. Cytoplasmic dynein plays a role in mammalian mitotic spindle formation. J. Cell Biol. 123: 849-858, 1993. [PubMed: 8227145] [Full Text: https://doi.org/10.1083/jcb.123.4.849]

  24. Vissers, L. E. L. M., de Ligt, J., Gilissen, C., Janssen, I., Steehouwer, M., de Vries, P., van Lier, B., Arts, P., Wieskamp, N., del Rosario, M., van Bon, B. W. M., Hoischen, A., de Vries, B. B. A., Brunner, H. G., Veltman, J. A. A de novo paradigm for mental retardation. Nature Genet. 42: 1109-1112, 2010. [PubMed: 21076407] [Full Text: https://doi.org/10.1038/ng.712]

  25. Weedon, M. N., Hastings, R., Caswell, R., Xie, W., Paszkiewicz, K., Antoniadi, T., Williams, M., King, C., Greenhalgh, L., Newbury-Ecob, R., Ellard, S. Exome sequencing identifies a DYNC1H1 mutation in a large pedigree with dominant axonal Charcot-Marie-Tooth disease. Am. J. Hum. Genet. 89: 308-312, 2011. [PubMed: 21820100] [Full Text: https://doi.org/10.1016/j.ajhg.2011.07.002]

  26. Willemsen, M. H., Vissers, L. E. L., Willemsen, M. A. A. P., van Bon, B. W. M., Kroes, T., de Ligt, J., de Vries, B. B., Schoots, J., Lugtenberg, D., Hamel, B. C. J., van Bokhoven, H., Brunner, H. G., Veltman, J. A., Kleefstra, T. Mutations in DYNC1H1 cause severe intellectual disability with neuronal migration defects. J. Med. Genet. 49: 179-183, 2012. [PubMed: 22368300] [Full Text: https://doi.org/10.1136/jmedgenet-2011-100542]


Contributors:
Ada Hamosh - updated : 04/16/2018
George E. Tiller - updated : 09/12/2017
Ada Hamosh - updated : 09/02/2014
Cassandra L. Kniffin - updated : 7/1/2014
Cassandra L. Kniffin - updated : 9/23/2013
Cassandra L. Kniffin - updated : 6/27/2013
Cassandra L. Kniffin - updated : 4/25/2012
Cassandra L. Kniffin - updated : 4/9/2012
Cassandra L. Kniffin - updated : 9/15/2011
Ada Hamosh - updated : 12/29/2008
Ada Hamosh - updated : 4/4/2008
Patricia A. Hartz - updated : 4/18/2006
Ada Hamosh - updated : 8/2/2005
Ada Hamosh - updated : 5/6/2003
Rebekah S. Rasooly - updated : 11/18/1998

Creation Date:
Victor A. McKusick : 9/13/1994

Edit History:
alopez : 04/07/2023
ckniffin : 04/07/2023
alopez : 04/01/2022
alopez : 04/01/2022
alopez : 04/16/2018
carol : 01/05/2018
alopez : 09/12/2017
alopez : 09/02/2014
carol : 7/2/2014
mcolton : 7/2/2014
ckniffin : 7/1/2014
carol : 9/26/2013
tpirozzi : 9/26/2013
ckniffin : 9/23/2013
carol : 8/28/2013
carol : 7/10/2013
carol : 7/9/2013
ckniffin : 6/27/2013
carol : 9/19/2012
terry : 7/5/2012
terry : 5/2/2012
carol : 4/27/2012
ckniffin : 4/25/2012
alopez : 4/10/2012
terry : 4/10/2012
terry : 4/10/2012
ckniffin : 4/9/2012
carol : 9/16/2011
ckniffin : 9/15/2011
alopez : 12/29/2008
terry : 12/29/2008
alopez : 4/11/2008
terry : 4/4/2008
mgross : 4/19/2006
mgross : 4/18/2006
alopez : 8/3/2005
terry : 8/2/2005
carol : 5/12/2004
alopez : 5/8/2003
terry : 5/6/2003
dkim : 12/18/1998
dkim : 12/3/1998
psherman : 12/2/1998
alopez : 11/18/1998
alopez : 8/21/1998
mimadm : 9/23/1995
carol : 9/13/1994